Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(7): 107432, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38825009

RESUMEN

The Ca2+-activated Cl- channel regulator CLCA1 potentiates the activity of the Ca2+-activated Cl- channel (CaCC) TMEM16A by directly engaging the channel at the cell surface, inhibiting its reinternalization and increasing Ca2+-dependent Cl- current (ICaCC) density. We now present evidence of functional pairing between two other CLCA and TMEM16 protein family members, namely CLCA4 and the CaCC TMEM16B. Similar to CLCA1, (i) CLCA4 is a self-cleaving metalloprotease, and the N-terminal portion (N-CLCA4) is secreted; (ii) the von Willebrand factor type A (VWA) domain in N-CLCA4 is sufficient to potentiate ICaCC in HEK293T cells; and (iii) this is mediated by the metal ion-dependent adhesion site motif within VWA. The results indicate that, despite the conserved regulatory mechanism and homology between CLCA1 and CLCA4, CLCA4-dependent ICaCC are carried by TMEM16B, rather than TMEM16A. Our findings show specificity in CLCA/TMEM16 interactions and suggest broad physiological and pathophysiological links between these two protein families.

2.
Curr Protoc ; 2(8): e512, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35998009

RESUMEN

Milligram quantities of pure proteins are required for structural, functional, and pharmaceutical screening studies. These requirements can be challenging for a majority of important therapeutic targets that are secreted glycoproteins, receptors, membrane proteins, or large cytosolic complexes. Here, we present protocols for producing and purifying large amounts of secreted glycoproteins using the mammalian cell-based Expi293F system via large-scale transient transfection. This system can be easily adapted for the production of membrane proteins and large cytosolic complexes. The method can be utilized to quickly evaluate numerous expression constructs to identify optimal expressers. Use of mammalian cells ensures proper post-translational modifications, including disulfide bonds and glycosylation, that can be important for accurate functional studies. In addition, minor modifications can be introduced to produce labeled or deglycosylated proteins for structural studies by X-ray crystallography, nuclear magnetic resonance, or cryo-electron microscopy. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Production of milligram quantities of plasmid DNA for large-scale transient transfection Basic Protocol 2: Large-scale culture and transient transfection of Expi293F cells Basic Protocol 3: Purification of hexahistidine-tagged proteins from medium.


Asunto(s)
Eucariontes , Glicoproteínas , Animales , Microscopía por Crioelectrón , Eucariontes/genética , Glicoproteínas/genética , Mamíferos/genética , Proteínas de la Membrana/genética , Transfección
3.
Cell ; 184(20): 5163-5178.e24, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34559985

RESUMEN

Rift Valley fever virus (RVFV) is a zoonotic pathogen with pandemic potential. RVFV entry is mediated by the viral glycoprotein (Gn), but host entry factors remain poorly defined. Our genome-wide CRISPR screen identified low-density lipoprotein receptor-related protein 1 (mouse Lrp1/human LRP1), heat shock protein (Grp94), and receptor-associated protein (RAP) as critical host factors for RVFV infection. RVFV Gn directly binds to specific Lrp1 clusters and is glycosylation independent. Exogenous addition of murine RAP domain 3 (mRAPD3) and anti-Lrp1 antibodies neutralizes RVFV infection in taxonomically diverse cell lines. Mice treated with mRAPD3 and infected with pathogenic RVFV are protected from disease and death. A mutant mRAPD3 that binds Lrp1 weakly failed to protect from RVFV infection. Together, these data support Lrp1 as a host entry factor for RVFV infection and define a new target to limit RVFV infections.


Asunto(s)
Interacciones Huésped-Patógeno , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Especificidad de Anticuerpos/inmunología , Secuencia de Bases , Encéfalo/patología , Encéfalo/virología , Sistemas CRISPR-Cas/genética , Membrana Celular/metabolismo , Células Cultivadas , Glicoproteínas/metabolismo , Glicosaminoglicanos/metabolismo , Glicosilación , Humanos , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/metabolismo , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Glicoproteínas de Membrana/metabolismo , Ratones , Unión Proteica , Desnaturalización Proteica , Fiebre del Valle del Rift/patología , Fiebre del Valle del Rift/prevención & control , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/inmunología
4.
Alzheimers Dement ; 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33090700

RESUMEN

INTRODUCTION: Triggering receptor expressed on myeloid cells-2 (TREM2) is an immune receptor expressed on microglia that also can become soluble (sTREM2). How TREM2 engages different ligands remains poorly understood. METHODS: We used comprehensive biolayer interferometry (BLI) analysis to investigate TREM2 and sTREM2 interactions with apolipoprotein E (apoE) and monomeric amyloid beta (Aß) (mAß42). RESULTS: TREM2 engagement of apoE was protein mediated with little effect of lipidation, showing slight affinity differences between isoforms (E4 > E3 > E2). Another family member, TREML2, did not bind apoE. Disease-linked TREM2 variants within a "basic patch" minimally impact apoE binding. Instead, TREM2 uses a unique hydrophobic surface to bind apoE, which requires the apoE hinge region. TREM2 and sTREM2 directly bind mAß42 and potently inhibit Aß42 polymerization, suggesting a potential role for soluble sTREM2 in preventing AD pathogenesis. DISCUSSION: These findings demonstrate that TREM2 has at least two ligand-binding surfaces that might be therapeutic targets and uncovers a potential function for sTREM2 in directly inhibiting Aß polymerization.

5.
Cell Rep ; 30(4): 1141-1151.e3, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995732

RESUMEN

The secreted protein calcium-activated chloride channel regulator 1 (CLCA1) utilizes a von Willebrand factor type A (VWA) domain to bind to and potentiate the calcium-activated chloride channel TMEM16A. To gain insight into this unique potentiation mechanism, we determined the 2.0-Å crystal structure of human CLCA1 VWA bound to Ca2+. The structure reveals the metal-ion-dependent adhesion site (MIDAS) in a high-affinity "open" conformation, engaging in crystal contacts that likely mimic how CLCA1 engages TMEM16A. The CLCA1 VWA contains a disulfide bond between α3 and α4 in close proximity to the MIDAS that is invariant in the CLCA family and unique in VWA structures. Further biophysical studies indicate that CLCA1 VWA is preferably stabilized by Mg2+ over Ca2+ and that α6 atypically extends from the VWA core. Finally, an analysis of TMEM16A structures suggests residues likely to mediate interaction with CLCA1 VWA.


Asunto(s)
Anoctamina-1/química , Anoctamina-1/metabolismo , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fenómenos Biofísicos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Dominios Proteicos , Pliegue de Proteína
6.
Biochemistry ; 58(6): 657-664, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30592210

RESUMEN

Members of Mononegavirales, the order that includes nonsegmented negative sense RNA viruses (NNSVs), encode a small number of multifunctional proteins. In members of the Filoviridae family, virus protein 35 (VP35) facilitates immune evasion and functions as an obligatory cofactor for viral RNA synthesis. VP35 functions in a manner orthologous to that of phosphoproteins from other NNSVs. Although the critical roles of Ebola viral VP35 (eVP35) in immune evasion and RNA synthesis are well-appreciated, a complete understanding of its organization and its role in carrying out its many functions has yet to be fully realized. In particular, we currently lack information about the role of the oligomerization domain within eVP35. To address this limitation, we report here an investigation of the oligomer structure of eVP35 using hybrid methods that include multiangle light scattering, small-angle X-ray scattering, and cross-linking coupled with mass spectrometry to determine the shape and orientation of the eVP35 oligomer. Our integrative results are consistent with a parallel tetramer in which the N-terminal regions that are required for RNA synthesis are all oriented in the same direction. Furthermore, these results define a framework for targeting the symmetric tetramer for structure-based antiviral discovery.


Asunto(s)
Proteínas Reguladoras y Accesorias Virales/química , Secuencia de Aminoácidos , Ebolavirus/química , Escherichia coli/genética , Dominios Proteicos , Multimerización de Proteína
7.
J Biol Chem ; 293(51): 19572-19585, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30355735

RESUMEN

Clinical isolates of Yersinia, Klebsiella, and Escherichia coli frequently secrete the small molecule metallophore yersiniabactin (Ybt), which passivates and scavenges transition metals during human infections. YbtT is encoded within the Ybt biosynthetic operon and is critical for full Ybt production in bacteria. However, its biosynthetic function has been unclear because it is not essential for Ybt production by the in vitro reconstituted nonribosomal peptide synthetase/polyketide synthase (NRPS/PKS) pathway. Here, we report the structural and biochemical characterization of YbtT. YbtT structures at 1.4-1.9 Å resolution possess a serine hydrolase catalytic triad and an associated substrate chamber with features similar to those previously reported for low-specificity type II thioesterases (TEIIs). We found that YbtT interacts with the two major Ybt biosynthetic proteins, HMWP1 (high-molecular-weight protein 1) and HMWP2 (high-molecular-weight protein 2), and hydrolyzes a variety of aromatic and acyl groups from their phosphopantetheinylated carrier protein domains. In vivo YbtT titration in uropathogenic E. coli revealed a distinct optimum for Ybt production consistent with a tradeoff between clearing both stalled inhibitory intermediates and productive Ybt precursors from HMWP1 and HMWP2. These results are consistent with a model in which YbtT maintains cellular Ybt biosynthesis by removing nonproductive, inhibitory thioesters that form aberrantly at multiple sites on HMWP1 and HMWP2.


Asunto(s)
Enterobacteriaceae/enzimología , Ácido Graso Sintasas/química , Ácido Graso Sintasas/metabolismo , Fenoles/metabolismo , Tiazoles/metabolismo , Tioléster Hidrolasas/química , Tioléster Hidrolasas/metabolismo , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Enterobacteriaceae/metabolismo , Ácido Graso Sintasas/genética , Hidrólisis , Cinética , Modelos Moleculares , Mutación , Tioléster Hidrolasas/genética
8.
Bioessays ; 40(10): e1800086, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30113067

RESUMEN

Recent findings regarding the cellular biology and immunology of BST-2 (also known as tetherin) indicate that its function could be exploited as a universal replication inhibitor of enveloped respiratory viruses (e.g., influenza, respiratory syncytial virus, etc.). BST-2 inhibits viral replication by preventing virus budding from the plasma membrane and by inducing an antiviral state in cells adjacent to infection via unique inflammatory signaling mechanisms. This review presents the first comprehensive summary of what is currently known about BST-2 anti-viral function against respiratory viruses, how these viruses construct countermeasures to antagonize BST-2, and how BST-2 function might be targeted to develop therapies to treat respiratory virus infections. The authors address the current gaps in knowledge, including the need for mechanistic understanding of BST-2 antagonism by respiratory viruses, that should be bridged to achieve that goal.


Asunto(s)
Antígenos CD/fisiología , Interacciones Huésped-Patógeno/fisiología , Infecciones del Sistema Respiratorio/virología , Antígenos CD/química , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/fisiología , Humanos , Terapia Molecular Dirigida/métodos , Transducción de Señal , Virión , Virosis/inmunología , Liberación del Virus , Replicación Viral/efectos de los fármacos
9.
J Mol Biol ; 429(11): 1607-1629, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28432014

RESUMEN

The protein triggering receptor expressed on myeloid cells-2 (TREM2) is an immunomodulatory receptor with a central role in myeloid cell activation and survival. In recent years, the importance of TREM2 has been highlighted by the identification of coding variants that increase risk for Alzheimer's disease and other neurodegenerative diseases. Animal studies have further shown the importance of TREM2 in neurodegenerative and other inflammatory disease models including chronic obstructive pulmonary disease, multiple sclerosis, and stroke. A mechanistic understanding of TREM2 function remains elusive, however, due in part to the absence of conclusive information regarding the identity of endogenous TREM2 ligands. While many TREM2 ligands have been proposed, their physiological role and mechanism of engagement remain to be determined. In this review, we highlight the suggested roles of TREM2 in these diseases and the recent advances in our understanding of TREM2 and discuss putative TREM2-ligand interactions and their potential roles in signaling during health and disease. We develop a model based on the TREM2 structure to explain how different TREM2 ligands might interact with the receptor and how disease risk variants may alter ligand interactions. Finally, we propose future experimental directions to establish the role and importance of these different interactions on TREM2 function.


Asunto(s)
Salud , Glicoproteínas de Membrana/metabolismo , Enfermedades Neurodegenerativas/patología , Receptores Inmunológicos/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Unión Proteica
10.
J Biol Chem ; 292(22): 9164-9174, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28420732

RESUMEN

Calcium-activated chloride channels (CaCCs) are key players in transepithelial ion transport and fluid secretion, smooth muscle constriction, neuronal excitability, and cell proliferation. The CaCC regulator 1 (CLCA1) modulates the activity of the CaCC TMEM16A/Anoctamin 1 (ANO1) by directly engaging the channel at the cell surface, but the exact mechanism is unknown. Here we demonstrate that the von Willebrand factor type A (VWA) domain within the cleaved CLCA1 N-terminal fragment is necessary and sufficient for this interaction. TMEM16A protein levels on the cell surface were increased in HEK293T cells transfected with CLCA1 constructs containing the VWA domain, and TMEM16A-like currents were activated. Similar currents were evoked in cells exposed to secreted VWA domain alone, and these currents were significantly knocked down by TMEM16A siRNA. VWA-dependent TMEM16A modulation was not modified by the S357N mutation, a VWA domain polymorphism associated with more severe meconium ileus in cystic fibrosis patients. VWA-activated currents were significantly reduced in the absence of extracellular Mg2+, and mutation of residues within the conserved metal ion-dependent adhesion site motif impaired the ability of VWA to potentiate TMEM16A activity, suggesting that CLCA1-TMEM16A interactions are Mg2+- and metal ion-dependent adhesion site-dependent. Increase in TMEM16A activity occurred within minutes of exposure to CLCA1 or after a short treatment with nocodazole, consistent with the hypothesis that CLCA1 stabilizes TMEM16A at the cell surface by preventing its internalization. Our study hints at the therapeutic potential of the selective activation of TMEM16A by the CLCA1 VWA domain in loss-of-function chloride channelopathies such as cystic fibrosis.


Asunto(s)
Canales de Cloruro/metabolismo , Magnesio/metabolismo , Mutación Missense , Proteínas de Neoplasias/metabolismo , Sustitución de Aminoácidos , Anoctamina-1 , Línea Celular , Canales de Cloruro/genética , Humanos , Proteínas de Neoplasias/genética , Dominios Proteicos , Estabilidad Proteica
11.
Biochim Biophys Acta ; 1860(11 Pt A): 2335-2344, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27369736

RESUMEN

BACKGROUND: P38 MAP kinases are centrally involved in mediating extracellular signaling in various diseases. While much attention has previously been focused on the ubiquitously expressed family member MAPK14 (p38α), recent studies indicate that family members such as MAPK13 (p38δ) display a more selective cellular and tissue expression and might therefore represent a specific kinase to target in certain diseases. METHODS: To facilitate the design of potent and specific inhibitors, we present here the structural, biophysical, and functional characterization of two new MAPK13-inhibitor complexes, as well as the first comprehensive structural, biophysical, and functional analysis of MAPK13 complexes with four different inhibitor compounds of greatly varying potency. RESULTS: These inhibitors display IC50 values either in the nanomolar range or micromolar range (>800-fold range). The nanomolar inhibitors exhibit much longer ligand-enzyme complex half-lives compared to the micromolar inhibitors as measured by biolayer interferometry. Crystal structures of the MAPK13 inhibitor complexes reveal that the nanomolar inhibitors engage MAPK13 in the DFG-out binding mode, while the micromolar inhibitors are in the DFG-in mode. Detailed structural and computational docking analyses suggest that this difference in binding mode engagement is driven by conformational restraints imposed by the chemical structure of the inhibitors, and may be fortified by an additional hydrogen bond to MAPK13 in the nanomolar inhibitors. CONCLUSIONS: These studies provide a structural basis for understanding the differences in potency exhibited by these inhibitors. GENERAL SIGNIFICANCE: They also provide the groundwork for future studies to improve specificity, potency, pharmacodynamics, and pharmacokinetic properties.


Asunto(s)
Proteína Quinasa 13 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Sitios de Unión , Humanos , Proteína Quinasa 13 Activada por Mitógenos/química , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad Cuantitativa
12.
Nat Commun ; 7: 10882, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26947396

RESUMEN

Access to experimental X-ray diffraction image data is fundamental for validation and reproduction of macromolecular models and indispensable for development of structural biology processing methods. Here, we established a diffraction data publication and dissemination system, Structural Biology Data Grid (SBDG; data.sbgrid.org), to preserve primary experimental data sets that support scientific publications. Data sets are accessible to researchers through a community driven data grid, which facilitates global data access. Our analysis of a pilot collection of crystallographic data sets demonstrates that the information archived by SBDG is sufficient to reprocess data to statistics that meet or exceed the quality of the original published structures. SBDG has extended its services to the entire community and is used to develop support for other types of biomedical data sets. It is anticipated that access to the experimental data sets will enhance the paradigm shift in the community towards a much more dynamic body of continuously improving data analysis.


Asunto(s)
Bases de Datos Genéticas , Sustancias Macromoleculares/química , Publicaciones , Cristalografía por Rayos X , Internet , Programas Informáticos
13.
Mediators Inflamm ; 2015: 497387, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26612971

RESUMEN

Chloride transport proteins play critical roles in inflammatory airway diseases, contributing to the detrimental aspects of mucus overproduction, mucus secretion, and airway constriction. However, they also play crucial roles in contributing to the innate immune properties of mucus and mucociliary clearance. In this review, we focus on the emerging novel roles for a chloride channel regulator (CLCA1), a calcium-activated chloride channel (TMEM16A), and two chloride exchangers (SLC26A4/pendrin and SLC26A9) in chronic inflammatory airway diseases.


Asunto(s)
Antiportadores/fisiología , Asma/etiología , Canales de Cloruro/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Anoctamina-1 , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Humanos , Factor de Transcripción STAT6/fisiología , Transportadores de Sulfato
14.
Nat Immunol ; 16(12): 1215-27, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26479788

RESUMEN

Enhancing the response to interferon could offer an immunological advantage to the host. In support of this concept, we used a modified form of the transcription factor STAT1 to achieve hyper-responsiveness to interferon without toxicity and markedly improve antiviral function in transgenic mice and transduced human cells. We found that the improvement depended on expression of a PARP9-DTX3L complex with distinct domains for interaction with STAT1 and for activity as an E3 ubiquitin ligase that acted on host histone H2BJ to promote interferon-stimulated gene expression and on viral 3C proteases to degrade these proteases via the immunoproteasome. Thus, PARP9-DTX3L acted on host and pathogen to achieve a double layer of immunity within a safe reserve in the interferon signaling pathway.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Histonas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Animales , Línea Celular , Núcleo Celular/metabolismo , Virus de la Encefalomiocarditis/fisiología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Interferón beta/farmacología , Interferón gamma/farmacología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Mutación , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Interferencia de ARN , ADN Polimerasa Dirigida por ARN , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Transcriptoma/efectos de los fármacos , Ubiquitina-Proteína Ligasas/genética
15.
Expert Rev Respir Med ; 9(5): 503-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26296094

RESUMEN

The hallmark traits of chronic obstructive airway diseases are inflammation, airway constriction due to hyperreactivity and mucus overproduction. The current common treatments for asthma and chronic obstructive pulmonary disease target the first two traits with none currently targeting mucus overproduction. The main source of obstructive mucus production is mucus cell metaplasia (MCM), the transdifferentiation of airway epithelial cells into mucus-producing goblet cells, in the small airways. Our current understanding of MCM is profusely incomplete. Few of the molecular players involved in driving MCM in humans have been identified and for many of those that have, their functions and mechanisms are unknown. This fact has limited the development of therapeutics that target mucus overproduction by inhibiting MCM. Current work in the field is aiming to change that.


Asunto(s)
Asma/fisiopatología , Canales de Cloruro/genética , Moco/fisiología , Proteínas de Neoplasias/genética , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Mucosa Respiratoria/fisiopatología , Anoctamina-1 , Asma/genética , Transdiferenciación Celular/genética , Células Epiteliales/fisiología , Humanos , Enfermedad Pulmonar Obstructiva Crónica/genética
16.
J Exp Med ; 212(5): 681-97, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25897174

RESUMEN

Viral infections and type 2 immune responses are thought to be critical for the development of chronic respiratory disease, but the link between these events needs to be better defined. Here, we study a mouse model in which infection with a mouse parainfluenza virus known as Sendai virus (SeV) leads to long-term activation of innate immune cells that drive IL-13-dependent lung disease. We find that chronic postviral disease (signified by formation of excess airway mucus and accumulation of M2-differentiating lung macrophages) requires macrophage expression of triggering receptor expressed on myeloid cells-2 (TREM-2). Analysis of mechanism shows that viral replication increases lung macrophage levels of intracellular and cell surface TREM-2, and this action prevents macrophage apoptosis that would otherwise occur during the acute illness (5-12 d after inoculation). However, the largest increases in TREM-2 levels are found as the soluble form (sTREM-2) long after clearance of infection (49 d after inoculation). At this time, IL-13 and the adapter protein DAP12 promote TREM-2 cleavage to sTREM-2 that is unexpectedly active in preventing macrophage apoptosis. The results thereby define an unprecedented mechanism for a feed-forward expansion of lung macrophages (with IL-13 production and consequent M2 differentiation) that further explains how acute infection leads to chronic inflammatory disease.


Asunto(s)
Apoptosis/inmunología , Enfermedades Pulmonares/inmunología , Macrófagos Alveolares/inmunología , Glicoproteínas de Membrana/inmunología , Receptores Inmunológicos/inmunología , Infecciones por Respirovirus/inmunología , Virus Sendai/fisiología , Animales , Apoptosis/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Inmunidad Innata/genética , Interleucina-13/genética , Interleucina-13/inmunología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/virología , Macrófagos Alveolares/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Receptores Inmunológicos/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/patología , Replicación Viral/genética , Replicación Viral/inmunología
17.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 790-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849390

RESUMEN

The p38 MAP kinases (p38 MAPKs) represent an important family centrally involved in mediating extracellular signaling. Recent studies indicate that family members such as MAPK13 (p38δ) display a selective cellular and tissue expression and are therefore involved in specific diseases. Detailed structural studies of all p38 MAPK family members are crucial for the design of specific inhibitors. In order to facilitate such ventures, the structure of MAPK13 was determined in both the inactive (unphosphorylated; MAPK13) and active (dual phosphorylated; MAPK13/pTpY) forms. Here, the first preparation, crystallization and structure determination of MAPK13/pTpY are presented and the structure is compared with the previously reported structure of MAPK13 in order to facilitate studies for structure-based drug design. A comprehensive analysis of inactive versus active structures for the p38 MAPK family is also presented. It is found that MAPK13 undergoes a larger interlobe configurational rearrangement upon activation compared with MAPK14. Surprisingly, the analysis of activated p38 MAPK structures (MAP12/pTpY, MAPK13/pTpY and MAPK14/pTpY) reveals that, despite a high degree of sequence similarity, different side chains are used to coordinate the phosphorylated residues. There are also differences in the rearrangement of the hinge region that occur in MAPK14 compared with MAPK13 which would affect inhibitor binding. A thorough examination of all of the active (phosphorylated) and inactive (unphosphorylated) p38 MAPK family member structures was performed to reveal a common structural basis of activation for the p38 MAP kinase family and to identify structural differences that may be exploited for developing family member-specific inhibitors.


Asunto(s)
Proteína Quinasa 13 Activada por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Activación Enzimática , Humanos , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Fosforilación , Conformación Proteica , Alineación de Secuencia , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Elife ; 42015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25781344

RESUMEN

Calcium-activated chloride channel regulator 1 (CLCA1) activates calcium-dependent chloride currents; neither the target, nor mechanism, is known. We demonstrate that secreted CLCA1 activates calcium-dependent chloride currents in HEK293T cells in a paracrine fashion, and endogenous TMEM16A/Anoctamin1 conducts the currents. Exposure to exogenous CLCA1 increases cell surface levels of TMEM16A and cellular binding experiments indicate CLCA1 engages TMEM16A on the surface of these cells. Altogether, our data suggest that CLCA1 stabilizes TMEM16A on the cell surface, thus increasing surface expression, which results in increased calcium-dependent chloride currents. Our results identify the first Cl(-) channel target of the CLCA family of proteins and establish CLCA1 as the first secreted direct modifier of TMEM16A activity, delineating a unique mechanism to increase currents. These results suggest cooperative roles for CLCA and TMEM16 proteins in influencing the physiology of multiple tissues, and the pathology of multiple diseases, including asthma, COPD, cystic fibrosis, and certain cancers.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/metabolismo , Activación del Canal Iónico , Proteínas de Neoplasias/metabolismo , Anoctamina-1 , Western Blotting , Calcio/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/farmacología , Cloruros/metabolismo , Cloruros/farmacología , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Microscopía Confocal , Proteínas de Neoplasias/genética , Comunicación Paracrina/efectos de los fármacos , Técnicas de Placa-Clamp , Unión Proteica , Interferencia de ARN
19.
Cell Host Microbe ; 16(2): 187-200, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25121748

RESUMEN

During antiviral defense, interferon (IFN) signaling triggers nuclear transport of tyrosine-phosphorylated STAT1 (PY-STAT1), which occurs via a subset of karyopherin alpha (KPNA) nuclear transporters. Many viruses, including Ebola virus, actively antagonize STAT1 signaling to counteract the antiviral effects of IFN. Ebola virus VP24 protein (eVP24) binds KPNA to inhibit PY-STAT1 nuclear transport and render cells refractory to IFNs. We describe the structure of human KPNA5 C terminus in complex with eVP24. In the complex, eVP24 recognizes a unique nonclassical nuclear localization signal (NLS) binding site on KPNA5 that is necessary for efficient PY-STAT1 nuclear transport. eVP24 binds KPNA5 with very high affinity to effectively compete with and inhibit PY-STAT1 nuclear transport. In contrast, eVP24 binding does not affect the transport of classical NLS cargo. Thus, eVP24 counters cell-intrinsic innate immunity by selectively targeting PY-STAT1 nuclear import while leaving the transport of other cargo that may be required for viral replication unaffected.


Asunto(s)
Ebolavirus/fisiología , Factor de Transcripción STAT1/metabolismo , Proteínas Virales/química , alfa Carioferinas/química , Transporte Activo de Núcleo Celular , Unión Competitiva , Núcleo Celular/metabolismo , Cristalografía por Rayos X , Células HEK293 , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Señales de Localización Nuclear , Fosfoproteínas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Virales/metabolismo
20.
Protein Expr Purif ; 96: 32-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24508568

RESUMEN

TREM-2 (triggering receptor expressed on myeloid cells-2) is an innate immune receptor expressed on dendritic cells, macrophages, osteoclasts, and microglia. Recent genetic studies have reported the occurrence of point mutations in TREM-2 that correlate with a dramatically increased risk for the development of neurodegenerative diseases, including Alzheimer's disease, frontotemporal dementia, and Parkinson's disease. Structural and biophysical studies of wild-type and mutant TREM-2 ectodomains are required to understand the functional consequences of these mutations. In order to facilitate these studies, we undertook the production and crystallization of these proteins. Here we demonstrate that, unlike many single Ig domain proteins, TREM-2 could not be readily refolded from bacterially-expressed inclusion bodies. Instead, we developed a mammalian-cell based expression system for the successful production of wild-type and mutant TREM-2 proteins in milligram quantities and a single-chromatography-step purification scheme that produced diffraction-quality crystals. These crystals diffract to a resolution of 3.3 Å and produce data sufficient for structure determination. We describe herein the procedures to produce wild-type and mutant human TREM-2 Ig domains in sufficient quantities for structural and biophysical studies. Such studies are crucial to understand the functional consequences of TREM-2 point mutations linked to the development of neurodegenerative diseases and, ultimately, to develop patient-specific molecular therapies to treat them.


Asunto(s)
Inflamación/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/ultraestructura , Enfermedades Neurodegenerativas/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/ultraestructura , Clonación Molecular , Cristalografía por Rayos X , Expresión Génica , Humanos , Glicoproteínas de Membrana/biosíntesis , Mutación , Pliegue de Proteína , Estructura Terciaria de Proteína , Receptores Inmunológicos/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA